Abstract
Epidemiological and pre-clinical studies support the anti-tumor effects of ω-3 PUFAs; however, the results from human trials are mixed, making it difficult to provide dietary guidelines or recommendations of ω-3 PUFAs for disease prevention or treatment. Understanding the molecular mechanisms by which ω-3 PUFAs inhibit cancer could lead to better nutritional paradigms and human trials to clarify their health effects. The ω-3 PUFAs exert their biological activities mainly through the formation of bioactive lipid metabolites. Here we discuss the biology of cyclooxygenase, lipoxygenase and cytochrome P450 enzymes-derived ω-3-series lipid metabolites on angiogenesis, inflammation and cancer.
Keywords: ω-3 polyunsaturated fatty acids, cyclooxygenase, lipoxygenase, cytochrome P450
Introduction
In the U.S., there are ~1,665,540 new cases and ~585,720 deaths from cancers expected in 2014. It is estimated that 30% of cancer in developed countries are diet-related1. Human studies support that dietary ω-3 polyunsaturated fatty acids (PUFAs), in particular eicosapentaenoic acid (EPA, 20:5ω-3) and docosahexaenoic acid (DHA, 22:6ω-3), may reduce cancer risks2–10; while ω-6 PUFAs, such as linoleic acid (18:2ω-6) and arachidonic acid (ARA, 20:4ω-6), could promote tumor progression11–16. For example, in the VITamins And Lifestyle (VITAL) Cohort, current use of fish oil, but not other dietary supplements, was associated with reduced risks of breast cancer10. The breast cancer patients with high tissue levels of DHA also respond better to chemotherapeutic drugs17. In contrary to the effects of ω-3 PUFAs, studies carried out in Mexico, Sweden, Singapore and China showed that dietary intake of ω-6 PUFAs was associated with increased breast cancer risks11, 14, 16, 18. This is important because current western diet contains 20–30 times more ω-6 than ω-3 PUFAs19. Validation of the anti-tumor effects of ω-3 PUFAs will have significant impact on public health. However, there are inconsistent results from human studies, which showed that ω-3 PUFAs had no effects20, 21 or detrimental effects on cancers22, making it difficult provide dietary guidelines or recommendations of ω-3 PUFAs for cancer prevention or treatment.
The inconsistent results could be due to many reasons. In previous ω-3 human trials, different types, doses and treatment durations of ω-3 PUFAs were used, making it difficult to compare and analyze the results of different ω-3 studies. Besides the heterogeneity of experimental designs, the mixed results could be, in part, due to inter-individual variations to metabolize ω-3 PUFAs to generate bioactive ω-3 lipid mediators (LMs)23–26. The ω-3 PUFAs act in part via formation of certain LMs such as cyclooxygenase (COX)-derived prostaglandin E3 (PGE3)27, lipoxygenase (LOX)-derived 4-hydroxy-docosahexaenoic acid (4-HDHA)28, cytochrome P450 (CYP)-derived epoxydocosapentaenoic acids (EDPs)29, as well as unique ω-3 lipid autacoids such as resolvins and protectins30, 31. These ω-3 LMs serve as autocrine and/or paracrine mediators to regulate inflammation and homeostasis32; many of these mediators are short-lived, locally produced and locally acting in response to cellular stimuli, followed with degradation or metabolism to maintain homeostasis33. The polymorphisms in the genes encoding ω-3 metabolism enzymes could affect the ω-3 metabolism, leading to different levels of ω-3/ω-6 LMs in tissues and varied biological responses to ω-3 supplementation. Indeed, recent research showed that there is a high degree of inter-individual variability in metabolizing ω-3 PUFAs to generate LMs upon dietary intake of ω-3 PUFAs23. Clearly, it is important to elucidate the specific lipid metabolizing enzymes and metabolites required for the anti-tumor effects of ω-3 PUFAs. The identified enzymes and metabolites could serve as biomarkers to screen the sub-populations which are most likely to respond to ω-3 PUFAs, or develop personalized doses for ω-3 supplementation. In addition, the bioactive ω-3 LMs could serve as biotemplates to design more potent and safer therapeutic drugs32.
The enzymatic metabolism of ω-6 ARA leads to formation of predominately though not exclusively pro-inflammatory and pro-tumorigenic LMs, which have been shown to play a central role in tumor progression34–36. Compared with the ω-6-series LMs, the roles of ω-3-series LMs in angiogenesis, inflammation and cancer are less known. The ω-3 LMs were thought to be less-active mediators, while emerging evidences support that certain classes of ω-3 LMs have potent effects to modulate inflammation, angiogenesis and tumorigenesis. In this review we will discuss the biology of COX, LOX and CYP-derived ω-3 LMs on angiogenesis, inflammation and cancer. The ω-3 PUFAs also act as precursors for biosynthesis of unique lipid autacoids such as resolvins and protectins, which also regulate multiple cellular processes including inflammation, angiogenesis and cancer31. These ω-3 autacoids have been discussed in several recent reviews30–32 and will not be discussed here.
ω-3 and ω-6 PUFAs
The ω-3 and ω-6 PUFAs are polyunsaturated fatty acids which have a double bond at the third and the sixth carbon atom from the end of the carbon chain respectively. Linoleic acid (LA, 18:2ω-6), which is an essential fatty acid and is highly abundant in common vegetable oils, is the major source of dietary ω-6 PUFA in the western diet. The average adult intake of LA in the U.S. ranges from 12–17 g/day for men and 9–11 g/day for women. LA can be further converted to arachidonic acid (ARA, 20:4ω-6), which is an important PUFA involved in cell signaling by generation of ω-6-series LMs (termed eicosanoids). Most research of ω-3 PUFAs have focused on EPA and DHA. Food sources of EPA and DHA include fish and fish oil supplements. Fish oil is among the most popular dietary supplements in United States. It is the most popular nonvitamin/nonmineral supplements in adults and the second most popular in children. In addition, major food companies are increasingly adding ω-3 PUFAs to various foods as value-added ingredients. FDA has approved Lovaza®, a mixture of EPA and DHA ethyl ester, as a prescription drug to treat hypertriglyceridemia (high levels of triglycerides). Another drug Vascepa® that is a pure EPA ethyl ester is currently seeking approval from FDA targeting hypertriglyceridemia. Recent technology development, using transgenic yeast, algae or supercritical carbon dioxide separation, allows the industry to prepare large-scale of highly purified EPA or DHA. Diets with a ω-6-to-ω-3 PUFA ratio of 1 are recommended by nutritionists, however, current western diets have a ratio of 20–30 due to too much consumption of LA and too low consumption of ω-3 PUFAs19. Due to the high dietary intake of ω-6 PUFAs, ARA is among the most abundant PUFAs in most tissues. The ω-3 PUFAs are highly enriched in retina and brain tissues, mainly in the form of DHA, the tissue levels of EPA are usually low37.
Molecular mechanisms of ω-3 PUFAs on cancer
Recent research shows that the lipid signaling is deregulated in tumor tissues, leading to increased production of pro-inflammatory and pro-tumorigenic eicosanoids from ARA in the tumor microenvironment. For example, the expressions of delta-6-desaturase which is a rate-limiting enzyme to convert linoleic acid to ARA, as well as phospholipases which release ARA from membrane phospholipids to initiate the biosynthesis of eicosanoids, are significantly up-regulated in tumor tissues38, 39. The expressions of pro-tumorigenic lipid metabolizing enzymes, such as COX-2, 5-LOX and CYP epoxygenases, have been reported to be up-regulated in tumors34, 36; while the anti-tumorigenic enzymes such as 15-LOX-1 are down-regulated34. Together, these changes lead to a supportive microenvironment to support tumor progression.
An important mechanism for the health-promoting effects of ω-3 PUFAs is that they suppress the metabolism of ARA to generate eicosanoids40. Upon dietary consumption, ω-3 PUFAs, including EPA and DHA, are incorporated into the membrane phospholipids at the expense of ω-6 ARA. Upon cellular stimulation, the incorporated ω-3 and ω-6 PUFAs are enzymatically released to generate intracellular free fatty acids (FFAs), which are rapidly metabolized by COX, LOX and CYP enzymes to generate ω-3-series and ω-6-series LMs. EPA and DHA inhibit the formation of ARA-derived ω-6-series LMs via multiple mechanisms, including reduced release of ARA from membrane phospholipids, inhibition of the enzymatic activities of the metabolizing enzymes, and direct competition with ARA for the enzymatic conversions. Besides inhibition of enzymatic metabolism of ARA, EPA and DHA also serve as alternative substrates of the lipid metabolism enzymes, leading to increased formation of ω-3-series LMs. Some of these mediators have potent effects to inhibit inflammation, angiogenesis and cancer28, 29, and will be discussed below.
COX-derived ω-3 LMs in angiogenesis, inflammation and cancer
The COX-2 pathway plays a critical role in angiogenesis, inflammation and cancer (Figure 1)34. The COX-2 metabolite of ARA, prostaglandin E2 (PGE2), is widely known to promote inflammation, neovascularization, primary tumor growth and metastasis. Increased expression of COX-2 has been observed in many tumor tissues34. COX-2 inhibitors, including non-steroidal anti-inflammatory drugs (NSAIDs) and cyclooxygenase (COX)-2 selective inhibitors (coxibs), have been shown to reduce cancer risks41, 42. However, the life-threatening cardiovascular risks as well as other adverse effects induced by long-term and high-dose use of these drugs have jeopardized their therapeutic applications43, 44.
Figure 1.
The ω-3 PUFAs reduce cancer risks via COX-2-dependent mechanisms. Human studies support that polymorphisms in the genes encoding COX-2 modulate the anti-tumor effects of ω-3 PUFAs45, 46. EPA and DHA reduced the formation of COX-2-derived PGE2, which contributes to the beneficial effects of ω-3 PUFAs40. DHA is widely believed not to be a substrate of COX enzymes, although it has been reported that DHA is converted by COX-2 to form hydroxyl DHA, which is further metabolized to generate electrophilic LMs with anti-inflammatory actions47. EPA has been shown to be an alternative substrate of COX-2, which converts it to the ω-3-series of prostaglandin termed prostaglandin E3 (PGE3) and other LMs. Compared with ARA, EPA is a poor substrate for COX enzymes48, 49. Previous studies have shown that PGE3 has less detrimental or even beneficial effects on cancer, the biology of PGE3 was discussed in a recent review27. PGE3 has been shown to have less pro-inflammatory and pro-angiogenic effects than PGE2. In NIH 3T3 fibroblasts, PGE2 induced cell proliferation while PGE3 had no effect in the same dose range. Both PGE2 and PGE3 induced COX-2 transcription in NIH 3T3 cells and IL-6 production in RAW 264.7 cells, but PGE3 had a significantly reduced pro-inflammatory effect50. In human endothelial cells, PGE2 further increased Ang2 expression induced by a combination of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), while PGE3 had no such effect51. PGE3 has also been shown to inhibit cancer cell proliferation and invasion. At a dose of 1 µM, PGE2 had no effect, while PGE3 inhibited proliferation of lung cancer A549 cells52. PGE3 also inhibited proliferation and induced apoptosis in B16F10 melanoma cells via mechanisms involving increased expression of PTEN53. In a Matrigel-based Boyden chamber assay, PGE3 inhibited cell invasion in the highly aggressive brain-metastatic melanoma 70W cell line54. The cellular receptors of PGE3 have not been confirmed; some studies have shown that PGE3 binds to the same receptors as PGE2 with reduced affinity and potency27, 55. Further, a recent study showed that Δ12-prostaglandin J3 (Δ12-PGJ3), which is a novel COX-derived metabolite of EPA, potently inhibited progression of leukemia in animal models56.
LOX-derived ω-3 LMs in angiogenesis, inflammation and cancer
The metabolism of PUFAs by LOX enzymes leads to the formation of leukotrienes and hydroxyl fatty acids (Figure 2)33. The LOX pathway in cancer is more complicated as there are multiple isoforms of LOX enzymes. It is generally believed that 5-LOX and 12-LOX and their metabolites promote cancer, while 15-LOX-1 and 15-LOX-2 have anti-tumor effects34, 57, 58. 5-HETE, a 5-LOX metabolite of ARA, has been shown to induce angiogenesis, inflammation, and tumor progression. Pharmacological inhibitors of the 5-LOX enzyme have been shown to suppress tumor progression in animal models34. Since both COX-2 and 5-LOX are up-regulated in tumor tissues, dual inhibition of COX-2 and 5-LOX has been shown to cause enhanced anti-tumor effect59, 60.
Figure 2.
The 5-LOX metabolites of ARA are generally believed to promote inflammation and angiogenesis34. Surprisingly, 5-LOX was recently shown to play a central role in the anti-angiogenic effect of DHA via formation of an anti-angiogenic metabolite 4-hydroxydocosahexaenoic acid (4-HDHA)28. Dietary supplementation of DHA has been shown to suppress retinal angiogenesis in an oxygen-induced retinopathy model61. Transgenic deletion of 5-LOX greatly reduced the anti-angiogenic effect of DHA, while deletion of COX-1/2 or 12/15-LOX had little effect, suggesting a central role of 5-LOX in the anti-angiogenic effect of DHA28. The 5-LOX enzyme mediates the anti-angiogenic effect of DHA via formation of 4-HDHA, which inhibited angiogenesis through a PPAR-γ-dependent mechanism28. Considering the importance of angiogenesis in tumor progression, it would be important to study the effect of the 4-HDHA pathway in tumor angiogenesis and associated tumor progression and metastasis.
EPA and DHA are also substrates of 15-LOX, which convert them to 15-hydroxyeicosapentaenoic acid (15-HEPE) and 17-hydroxy-docosahexaenoic acid (17-HDHA) respectively62, 63. Both 15-HEPE and 17-HDHA have been shown to inhibit the enzymatic activity of 5-LOX (a major enzyme to generate pro-inflammatory LMs), suggesting their potential anti-inflammatory effects62, 64. Further animal studies demonstrate the potent anti-inflammatory effects of 17-HDHA in colitis models. In a dextran sulfate sodium (DSS)-or 2,4,6-trinitrobenzene sulfonic acid-induced colitis model, treatment with 0.1–1 µg/animal/day 17-HDHA significantly reduced the disease activity index, body weight loss, colonic damage and polymorphonuclear infiltration in both colitis models. 17-HDHA also reduced levels of pro-inflammatory cytokines such as TNF-α, IL-1β, MIP-2, and CXCL1/KC and mRNA expression of NF-κB and adhesion molecules in colon tissue65. In another study, 17-HDHA also suppressed DSS-induced colitis in mice. In murine macrophage RAW264.7 cells, 17-HDHA increased phagocytosis in macrophages and promoted polarization towards an anti-inflammatory M2 phenotype66. These studies demonstrate the potent anti-inflammatory effect of 17-HDHA, indicating a critical role of the 15-LOX enzyme in the biological activities of DHA. 17-HDHA is a precursor for the biosynthesis of resolvins, also shown to have potent anti-inflammatory effects in recent reviews25, 26.
The 15-LOX metabolites of EPA and DHA have also been shown to directly inhibit cancer cell proliferation. EPA-derived 15-HEPE inhibited the formation of PGE2 and 5-HETE, as well as cancer cell proliferation in PC-3 and LNCaP cells67. DHA-derived 17-HDHA inhibited the proliferation of prostate cancer cells (PC-3, LNCaP and DU145) at doses much lower than the corresponding metabolite of ARA (15-HETE) and DHA68. The other 15-LOX metabolites of DHA, including 17-hydroperoxy-, 10,17-dihydroxy- and 7,17-dihydroxy-DHA, also inhibited cell proliferation via mechanisms involving activation of PPAR-γ and syndecan-1 signaling in prostate cancer cells68. The 15-LOX-mediated metabolism is required for the effect of DHA to induce syndecan-1 signaling and apoptosis in prostate cancer cells69. In another study, 17-hydroperoxy-DHA inhibited cell proliferation in neuroblastoma cells with an IC50 of 3–6 µM, compared with 12–15 µM for DHA70.
Human studies support a critical role of LOX in the effects of ω-3 PUFAs. Carriers of variant 5-LOX genotypes have been shown to have increased risks for inflammation and atherosclerosis compared with the carriers of common allele. Dietary intake of ω-6 PUFAs increased, while intake of ω-3 PUFAs decreased, the risk of atherosclerosis only in the carriers of variant alleles but not in the common alleles71. This study suggests that 5-LOX may be a biomarker to distinguish people who respond to the anti-atherosclerotic benefits of ω-3 PUFAs from non-responders71. In terms of cancer, Wang et al. showed that there is a significant interaction of the polymorphism in the genes encoding 5-LOX-activating protein (ALOX5AP) and dietary intake of ω-6 PUFA LA in terms of breast cancer risks in a population-based case-control study in San Francisco bay area12. Among the women with high dietary intake of LA, carrying the ALOX5AP −4900 AA genotype was associated with higher risk of breast cancer compared with other genotypes. No such correlations were observed in women consuming low levels of LA12. It would be interesting to test whether dietary intervention with ω-3 PUFAs would inhibit breast cancer progression in this high-risk sub-population. These two human studies demonstrate a strong diet-gene interaction, supporting the critical importance of PUFA metabolism enzyme in the biological activity of ω-3 and ω-6 PUFAs.
CYP-derived ω-3 LMs in angiogenesis, inflammation and cancer
The CYP pathway has two branches, converting ARA to epoxyeicosatrienoic oacids (EETs) by CYP epoxygenases (largely CYP2C and CYP2J) and 20-hydroxyeicosatetraenoic acid (20-HETE) by CYP ω-hydroxylase (largely CYP4A and CYP4F)72. EETs have been shown to have an array of beneficial actions, including anti-inflammatory, vasodilatory, anti-hypertensive, renal-protective, cardio-protective, and tissue regenerative actions. Pharmacological inhibitors of soluble epoxide hydrolase (sEH, the major enzyme to degrade EETs) which stabilize and increase EETs, are being developed to treat many human disorders72. 20-HETE has been shown to have predominately detrimental effects, inducing inflammation, vasoconstriction, hypertension, and cardiovascular problems. Pharmacological inhibitors of 20-HETE biosynthesis have been shown to alleviate many disease states in animal models73.
The roles of EETs and 20-HETE in cancer have not been well studied. EETs are mildly pro-angiogenic, stimulating endothelial cell proliferation, migration, invasion and tube formation72, as well as tissue regeneration74. Due to the pro-angiogenic actions, increased levels of EETs stimulate primary tumor growth and metastasis in implantable and spontaneous transgenic murine tumor models75. On the other hand, EETs are anti-inflammatory, where increased level of EETs inhibited colon cancer progression and tumor-associated inflammation in inflammation-driven colon cancer models76, 77. 20-HETE has been shown to increase primary tumor growth in murine tumor models, in part via induction of tumor angiogenesis and inflammation. Pharmacological inhibitors targeting 20-HETE biosynthesis have been shown to inhibit tumor progression in animal models35.
EPA and DHA have been shown to be highly efficient alternative substrates of CYP epoxygenases, leading to the formation of epoxygenated ω-3 PUFAs termed epoxyeicosatetraenoic acids (EEQs) and epoxydocosapentaenoic acids (EDPs) respectively72, 78. CYP epoxygenases selectively catalyze the epoxidation of the terminal double bond of ω-3 PUFAs, leading to the predominate formation of 17,18-EEQ from EPA and 19,20-EDP from DHA78–81. Compared with other DHA epoxide regioisomers, 19,20-EDP is the poorest substrate of sEH, which increases the relative proportion of 19,20-EDP in tissues82. Many studies have shown that ω-3 supplementation significantly increased levels of EEQs and EDPs in animal and human plasma and tissues78, 83–87.
EEQs and EDPs have similar or more potent effects for vasodilation, anti-inflammation and analgesia than EETs. EPA-derived 17,18-EEQ, as well as ARA-derived 14–15-EET, inhibited TNF-α-induced inflammation in human bronchi via NF-κB- and PPAR-γ-related mechanisms88, 89. In a carrageenan-induced inflammatory pain model in rats, all epoxygenated PUFAs (EETs, EEQs and EDPs) inhibited inflammatory pain, while the effects of EEQs were less potent than those of EETs and EDPs82. Also, a 12-LOX-derived metabolite of 17,18-EEQ, 12- hydroxy-17,18-epoxyeicosatetraenoic acid (12-OH-17,18-EEQ), inhibited LTB4-induced neutrophil chemotaxis and polarization in vitro at a low nM range90. In terms of vasodilation, EDPs are among the most potent vasodilators ever discovered (dilation EC50 = 0.5–24 pM)91. Direct treatment of EDPs suppressed Angiotensin II-induced hypertension in mice92. The CYP-mediated formation of EDPs has been hypothesized to contribute to the anti-hypertensive effects of DHA, as shown by transgenic deletion of CYP1A1 (a CYP epoxygenase enzyme) which attenuated the anti-hypertensive effects of DHA93.
Opposite to the pro-angiogenic effects of EETs, the ω-3-series fatty acid epoxides (EEQs and EDPs) have been shown to inhibit angiogenesis. 17,18-EEQ, but not other EEQ regioisomers, inhibited cell proliferation in the immortalized endothelial cell line bEND.3 at a dose of 10 µM, while EETs at the same dose range showed opposite effects to increase cell proliferation in bEND.3 cells94. This study suggests a potential anti-angiogenic effect of EEQs, however, more studies are needed to characterize their effects on angiogenesis, in particular in animal models of neovasculization. Our recent study showed that EDPs potently inhibited angiogenesis, primary tumor growth and metastasis29. In a Matrigel plug assay in mice, all EDP regioisomers (except 4,5-EDP which is chemically unstable) inhibited VGEF-induced angiogenesis. 19,20-EDP, which is a major EDP isomer in tissues, inhibited VEGF-induced angiogenesis with an EC50 value of 0.3 µg/animal, suggesting its potent anti-angiogenic effect. 19,20-EDP also suppressed basic fibroblast growth factor (bFGF)-induced angiogenesis in mice, suggesting a potential broad-spectrum anti-angiogenic effect. In human endothelial cells, 19,20-EDP inhibited endothelial tube formation, migration, and production of matrix metalloproteinases, via a mechanism involving VEGF receptor 2 (VEGFR2)-dependent signaling. Given that tumor metastasis causes 90% of human cancer deaths, anti-metastatic agents are very important therapeutic agents95. We demonstrated that two EDP regioisomers (i.e.,16,17-EDP and 19,20-EDP, dose = 0.05 mg/kg/day), when stabilized in circulation by co-administration of a selective sEH inhibitor, suppressed ~70% of tumor metastasis in mice29. In fact, EDPs are the first fatty acid metabolites to be shown to have anti-metastatic activities. Moreover, the stabilized EDP also inhibited Met-1 breast tumor growth (a highly aggressive triple-negative breast cancer model) in mice by ~70%29. Our findings demonstrate potent effects of EDPs on tumor angiogenesis, however, two recent studies showed that EDPs did not impact angiogenesis in retinal angiogenesis models96, 97. More studies are needed to characterize the effects and mechanisms of ω-3-series epoxides and diols on angiogenesis in different disease models as it is likely that the effects of these LMs may be disease- and tissue-specific.
Future Directions
The ω-3 PUFAs are among the most intensively studied nutritional compounds, as demonstrated by epidemiological and pre-clinical studies. However, after decades of ω-3 PUFA research, many of the health claims of ω-3 PUFAs remain controversial and have therefore had limited impact in disease prevention and treatment. The mixed results obtained with the use of ω-3 PUFAs in human trials may result in part from failing to recognize the importance of ω-3 PUFA metabolism. As we have discussed in this review, the enzymatic metabolism of ω-3 PUAFs generates ω-3-series LMs, which have potent actions to regulate inflammation, angiogenesis and tumor progression. The ω-3 LMs, rather than the parent ω-3 PUFAs (EPA or DHA), are more likely to be the ultimate bioacitve species interacting with cellular targets to exert the biological effects of ω-3 PUFA supplementation. However, the vast majority of previous ω-3 PUFA research has focused on tissue levels of ω-3 PUFAs, instead of ω-3 LMs, as biomarkers to establish the nutritional or therapeutic effects of ω-3 PUFAs.
As discussed above, it is critical to elucidate the lipid metabolizing enzymes and metabolites which are required for the biological effects of ω-3 PUFAs. We expect that increased dietary intake of ω-3 PUFAs is associated with reduced cancer risks among those with genetic variant that result in increased activity of the required ω-3 metabolizing enzymes. Recently, the development of transgenic animal models, LC-MS/MS-based lipidomics and standards for LMs has greatly facilitated the study of LMs. In the past decade, transgenic animal models with deletion or over-expression of lipid metabolism enzymes (COX-2, COX-1, 5-LOX, 12/15-LOX, CYP epoxygenase, CYP ω-hydroxylase, sEH, etc) have been developed and many of them are commercially available. Multiple laboratories in the U.S. have developed LC-MS/MS-based lipidomics methods, which can systematically analyze >100 LMs derived from ω-3/ω-6 PUFAs using minimal plasma or tissues98, 99. Many ω-3/ω-6 LM have been chemically synthesized and some of these LMs are commercially available, allowing cell culture and animal experiments to directly study their effects and mechanisms. These resources will greatly help to elucidate the roles of specific lipid metabolism pathway(s) and metabolite(s) in the effects of ω-3 and ω-6 PUFAs in different disease states. The knowledge obtained in pre-clinical models must be further verified in human trials. The identified lipid metabolism enzymes or metabolites could be used as biomarkers to distinguish "ω-3 responders" from "non-responders", leading to targeted human trials. Such knowledge could also help to provide personalized dietary recommendations. For example, sub-populations carrying certain genotypes of 5-LOX or 5-LOXAP could be educated to optimize their diet12, 71. An in-depth understanding of the molecular mechanisms of PUFAs, together with utilization of nutrigenomic and metabolomic approaches, could lead to targeted nutritional paradigms to better understand the metabolic individuality and nutrition effects of ω-3 PUFAs on human health100.
Figure 3.
Acknowledgement
We acknowledge support from R01 ES02710 and NIEHS Superfund P42 ES04699 (to B.D.H.), R01 CA134659, R01 CA112356, R01 CA103828 and Research Investments in the Sciences and Engineering (RISE) Program of UC Davis (to K.W.F.), R01 CA148633 (to D.P.), and Armstrong Fund of Science Award of UMass Amherst (to G.Z.). B.D.H. is a George and Judy Marcus Senior Fellow of the American Asthma Society.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Key TJ, Allen NE, Spencer EA, Travis RC. The effect of diet on risk of cancer. Lancet. 2002;360(9336):861–868. doi: 10.1016/S0140-6736(02)09958-0. [DOI] [PubMed] [Google Scholar]
- 2.Sasazuki S, Inoue M, Iwasaki M, et al. Intake of n-3 and n-6 polyunsaturated fatty acids and development of colorectal cancer by subsite: Japan Public Health Center-based prospective study. Int J Cancer. 2011;129(7):1718–1729. doi: 10.1002/ijc.25802. [DOI] [PubMed] [Google Scholar]
- 3.Murff HJ, Shrubsole MJ, Cai Q, et al. Dietary intake of PUFAs and colorectal polyp risk. Am J Clin Nutr. 2012;95(3):703–712. doi: 10.3945/ajcn.111.024000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kim S, Sandler DP, Galanko J, Martin C, Sandler RS. Intake of polyunsaturated fatty acids and distal large bowel cancer risk in whites and African Americans. Am J Epidemiol. 2010;171(9):969–979. doi: 10.1093/aje/kwq032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hall MN, Chavarro JE, Lee IM, Willett WC, Ma J. A 22-year prospective study of fish, n-3 fatty acid intake, and colorectal cancer risk in men. Cancer Epidemiol Biomarkers Prev. 2008;17(5):1136–1143. doi: 10.1158/1055-9965.EPI-07-2803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Almendingen K, Hostmark AT, Fausa O, Mosdol A, Aabakken L, Vatn MH. Familial adenomatous polyposis patients have high levels of arachidonic acid and docosahexaenoic acid and low levels of linoleic acid and alpha-linolenic acid in serum phospholipids. Int J Cancer. 2007;120(3):632–637. doi: 10.1002/ijc.22337. [DOI] [PubMed] [Google Scholar]
- 7.Schloss I, Kidd MS, Tichelaar HY, Young GO, O'Keefe SJ. Dietary factors associated with a low risk of colon cancer in coloured west coast fishermen. S Afr Med J. 1997;87(2):152–158. [PubMed] [Google Scholar]
- 8.Pot GK, Geelen A, van Heijningen EM, Siezen CL, van Kranen HJ, Kampman E. Opposing associations of serum n-3 and n-6 polyunsaturated fatty acids with colorectal adenoma risk: an endoscopy-based case-control study. Int J Cancer. 2008;123(8):1974–1977. doi: 10.1002/ijc.23729. [DOI] [PubMed] [Google Scholar]
- 9.Wolk A, Larsson SC, Johansson JE, Ekman P. Long-term fatty fish consumption and renal cell carcinoma incidence in women. JAMA. 2006;296(11):1371–1376. doi: 10.1001/jama.296.11.1371. [DOI] [PubMed] [Google Scholar]
- 10.Brasky TM, Lampe JW, Potter JD, Patterson RE, White E. Specialty supplements and breast cancer risk in the VITamins And Lifestyle (VITAL) Cohort. Cancer Epidemiol Biomarkers Prev. 2010;19(7):1696–1708. doi: 10.1158/1055-9965.EPI-10-0318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chajes V, Torres-Mejia G, Biessy C, et al. omega-3 and omega-6 polyunsaturated fatty acid intakes and the risk of breast cancer in Mexican women: impact of obesity status. Cancer Epidemiol Biomarkers Prev. 2012;21:319–326. doi: 10.1158/1055-9965.EPI-11-0896. [DOI] [PubMed] [Google Scholar]
- 12.Wang J, John E, Ingles S. 5-lipoxygenase and 5-lipoxygenase-activating protein gene polymorphisms, dietary linoleic acid, and risk for breast cancer. Cancer Epidemiol Biomarkers Prev. 2008;17:2748–2754. doi: 10.1158/1055-9965.EPI-08-0439. [DOI] [PubMed] [Google Scholar]
- 13.Thiebaut A, Chajes V, Gerber M, et al. Dietary intakes of omega-6 and omega-3 polyunsaturated fatty acids and the risk of breast cancer. Int J Cancer. 2009;124:924–931. doi: 10.1002/ijc.23980. [DOI] [PubMed] [Google Scholar]
- 14.Sonestedt E, Ericson U, Gullberg B, Skog K, Olsson H, Wirfalt E. Do both heterocyclic amines and omega-6 polyunsaturated fatty acids contribute to the incidence of breast cancer in postmenopausal women of the Malmo diet and cancer cohort? Int J Cancer. 2008;123:1637–1643. doi: 10.1002/ijc.23394. [DOI] [PubMed] [Google Scholar]
- 15.Murff H, Shu X, Li H, et al. Dietary polyunsaturated fatty acids and breast cancer risk in Chinese women: a prospective cohort study. Int J Cancer. 2011;128:1434–1441. doi: 10.1002/ijc.25703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gago-Dominguez M, Yuan J, Sun C, Lee H, Yu M. Opposing effects of dietary n-3 and n-6 fatty acids on mammary carcinogenesis: The Singapore Chinese Health Study. Br J Cancer. 2003;89:1686–1692. doi: 10.1038/sj.bjc.6601340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bougnoux P, Hajjaji N, Maheo K, Couet C, Chevalier S. Fatty acids and breast cancer: sensitization to treatments and prevention of metastatic re-growth. Prog Lipid Res. 2010;49(1):76–86. doi: 10.1016/j.plipres.2009.08.003. [DOI] [PubMed] [Google Scholar]
- 18.Murff HJ, Shu XO, Li H, et al. Dietary polyunsaturated fatty acids and breast cancer risk in Chinese women: a prospective cohort study. Int J Cancer. 2011;128(6):1434–1441. doi: 10.1002/ijc.25703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Simopoulos AP. Essential fatty acids in health and chronic disease. The American Journal of Clinical Nutrition. 1999;70(3):560s–569s. doi: 10.1093/ajcn/70.3.560s. [DOI] [PubMed] [Google Scholar]
- 20.Kobayashi M, Tsubono Y, Otani T, Hanaoka T, Sobue T, Tsugane S. Fish, long-chain n-3 polyunsaturated fatty acids, and risk of colorectal cancer in middle-aged Japanese: the JPHC study. Nutr Cancer. 2004;49(1):32–40. doi: 10.1207/s15327914nc4901_5. [DOI] [PubMed] [Google Scholar]
- 21.Akedo I, Ishikawa H, Nakamura T, et al. Three Cases with Familial Adenomatous Polyposis Diagnosed as Having Malignant Lesions in the Course of a Long-Term Trial Using Docosahexanoic Acid (DHA)-Concentrated Fish Oil Capsules. Japanese Journal of Clinical Oncology. 1998;28(12):762–765. doi: 10.1093/jjco/28.12.762. [DOI] [PubMed] [Google Scholar]
- 22.Stern MC, Butler LM, Corral R, et al. Polyunsaturated fatty acids, DNA repair single nucleotide polymorphisms and colorectal cancer in the Singapore Chinese Health Study. J Nutrigenet Nutrigenomics. 2009;2(6):273–279. doi: 10.1159/000308467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Nording ML, Yang J, Georgi K, et al. Individual variation in lipidomic profiles of healthy subjects in response to omega-3 Fatty acids. PLoS One. 2013;8(10):e76575. doi: 10.1371/journal.pone.0076575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Simopoulos AP. Genetic variants in the metabolism of omega-6 and omega-3 fatty acids: their role in the determination of nutritional requirements and chronic disease risk. Exp Biol Med (Maywood) 2010;235(7):785–795. doi: 10.1258/ebm.2010.009298. [DOI] [PubMed] [Google Scholar]
- 25.Stephensen CB, Armstrong P, Newman JW, et al. ALOX5 gene variants affect eicosanoid production and response to fish oil supplementation. J Lipid Res. 2011;52(5):991–1003. doi: 10.1194/jlr.P012864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Rudkowska I, Paradis A-M, Thifault E, et al. Differences in metabolomic and transcriptomic profiles between responders and non-responders to an n-3 polyunsaturated fatty acids (PUFAs) supplementation. Genes & Nutrition. 2013;8(4):411–423. doi: 10.1007/s12263-012-0328-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Yang P, Jiang Y, Fischer SM. Prostaglandin E3 metabolism and cancer. Cancer Lett. 2014 doi: 10.1016/j.canlet.2014.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sapieha P, Stahl A, Chen J, et al. 5-Lipoxygenase metabolite 4-HDHA is a mediator of the antiangiogenic effect of omega-3 polyunsaturated fatty acids. Sci Transl Med. 2011;3(69):69ra12. doi: 10.1126/scitranslmed.3001571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zhang G, Panigrahy D, Mahakian LM, et al. Epoxy metabolites of docosahexaenoic acid (DHA) inhibit angiogenesis, tumor growth, and metastasis. Proc Natl Acad Sci U S A. 2013;110(16):6530–6535. doi: 10.1073/pnas.1304321110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Serhan CN. Novel lipid mediators and resolution mechanisms in acute inflammation: to resolve or not? Am J Pathol. 2010;177(4):1576–1591. doi: 10.2353/ajpath.2010.100322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Serhan CN, Petasis NA. Resolvins and protectins in inflammation resolution. Chem Rev. 2011;111(10):5922–5943. doi: 10.1021/cr100396c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature. 2014;510(7503):92–101. doi: 10.1038/nature13479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Funk CD. Prostaglandins and leukotrienes: advances in eicosanoid biology. Science. 2001;294(5548):1871–1875. doi: 10.1126/science.294.5548.1871. [DOI] [PubMed] [Google Scholar]
- 34.Wang D, Dubois RN. Eicosanoids and cancer. Nat Rev Cancer. 2010;10(3):181–193. doi: 10.1038/nrc2809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Panigrahy D, Kaipainen A, Greene ER, Huang S. Cytochrome P450-derived eicosanoids: the neglected pathway in cancer. Cancer Metastasis Rev. 2010;29(4):723–735. doi: 10.1007/s10555-010-9264-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Panigrahy D, Greene ER, Pozzi A, Wang DW, Zeldin DC. EET signaling in cancer. Cancer Metastasis Rev. 2011;30(3–4):525–540. doi: 10.1007/s10555-011-9315-y. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 37.Arterburn LM, Hall EB, Oken H. Distribution, interconversion, and dose response of n-3 fatty acids in humans. Am J Clin Nutr. 2006;83(6) Suppl:1467S–1476S. doi: 10.1093/ajcn/83.6.1467S. [DOI] [PubMed] [Google Scholar]
- 38.Pender-Cudlip MC, Krag KJ, Martini D, et al. Delta-6-desaturase activity and arachidonic acid synthesis are increased in human breast cancer tissue. Cancer Sci. 2013;104(6):760–764. doi: 10.1111/cas.12129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Graff JR, Konicek BW, Deddens JA, et al. Expression of Group IIa Secretory Phospholipase A2 Increases with Prostate Tumor Grade. Clinical Cancer Research. 2001;7(12):3857–3861. [PubMed] [Google Scholar]
- 40.Rose DP, Connolly JM. Omega-3 fatty acids as cancer chemopreventive agents. Pharmacol Ther. 1999;83(3):217–244. doi: 10.1016/s0163-7258(99)00026-1. [DOI] [PubMed] [Google Scholar]
- 41.Farrow DC, Vaughan TL, Hansten PD, et al. Use of aspirin and other nonsteroidal anti-inflammatory drugs and risk of esophageal and gastric cancer. Cancer Epidemiol Biomarkers Prev. 1998;7(2):97–102. [PubMed] [Google Scholar]
- 42.Gupta RA, DuBois RN. Colorectal cancer prevention and treatment by inhibition of cyclooxygenase-2. Nat Rev Cancer. 2001;1(1):11–21. doi: 10.1038/35094017. [DOI] [PubMed] [Google Scholar]
- 43.Bresalier RS, Sandler RS, Quan H, et al. Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N Engl J Med. 2005;352(11):1092–1102. doi: 10.1056/NEJMoa050493. [DOI] [PubMed] [Google Scholar]
- 44.FitzGerald GA. Coxibs and cardiovascular disease. N Engl J Med. 2004;351(17):1709–1711. doi: 10.1056/NEJMp048288. [DOI] [PubMed] [Google Scholar]
- 45.Hedelin M, Chang ET, Wiklund F, et al. Association of frequent consumption of fatty fish with prostate cancer risk is modified by COX-2 polymorphism. Int J Cancer. 2007;120(2):398–405. doi: 10.1002/ijc.22319. [DOI] [PubMed] [Google Scholar]
- 46.Fradet V, Cheng I, Casey G, Witte JS. Dietary omega-3 fatty acids, cyclooxygenase-2 genetic variation, and aggressive prostate cancer risk. Clin Cancer Res. 2009;15(7):2559–2566. doi: 10.1158/1078-0432.CCR-08-2503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Groeger AL, Cipollina C, Cole MP, et al. Cyclooxygenase-2 generates anti-inflammatory mediators from omega-3 fatty acids. Nat Chem Biol. 2010;6(6):433–441. doi: 10.1038/nchembio.367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Malkowski MG, Thuresson ED, Lakkides KM, et al. Structure of eicosapentaenoic and linoleic acids in the cyclooxygenase site of prostaglandin endoperoxide H synthase-1. J Biol Chem. 2001;276(40):37547–37555. doi: 10.1074/jbc.M105982200. [DOI] [PubMed] [Google Scholar]
- 49.Laneuville O, Breuer DK, Xu N, et al. Fatty Acid Substrate Specificities of Human Prostaglandin-endoperoxide H Synthase-1 and −2: FORMATION OF 12-HYDROXY-(9Z,13E/Z,15Z)-OCTADECATRIENOIC ACIDS FROM α-LINOLENIC ACID. J Bio Chem. 1995;270(33):19330–19336. doi: 10.1074/jbc.270.33.19330. [DOI] [PubMed] [Google Scholar]
- 50.Bagga D, Wang L, Farias-Eisner R, Glaspy JA, Reddy ST. Differential effects of prostaglandin derived from omega-6 and omega-3 polyunsaturated fatty acids on COX-2 expression and IL-6 secretion. Proc Natl Acad Sci U S A. 2003;100(4):1751–1756. doi: 10.1073/pnas.0334211100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Szymczak M, Murray M, Petrovic N. Modulation of angiogenesis by omega-3 polyunsaturated fatty acids is mediated by cyclooxygenases. Blood. 2008;111(7):3514–3521. doi: 10.1182/blood-2007-08-109934. [DOI] [PubMed] [Google Scholar]
- 52.Yang P, Chan D, Felix E, et al. Formation and antiproliferative effect of prostaglandin E(3) from eicosapentaenoic acid in human lung cancer cells. J Lipid Res. 2004;45(6):1030–1039. doi: 10.1194/jlr.M300455-JLR200. [DOI] [PubMed] [Google Scholar]
- 53.Xia S, Lu Y, Wang J, et al. Melanoma growth is reduced in fat-1 transgenic mice: impact of omega-6/omega-3 essential fatty acids. Proc Natl Acad Sci U S A. 2006;103(33):12499–12504. doi: 10.1073/pnas.0605394103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Denkins Y, Kempf D, Ferniz M, Nileshwar S, Marchetti D. Role of omega-3 polyunsaturated fatty acids on cyclooxygenase-2 metabolism in brain-metastatic melanoma. J Lipid Res. 2005;46(6):1278–1284. doi: 10.1194/jlr.M400474-JLR200. [DOI] [PubMed] [Google Scholar]
- 55.Hawcroft G, Loadman PM, Belluzzi A, Hull MA. Effect of eicosapentaenoic acid on E-type prostaglandin synthesis and EP4 receptor signaling in human colorectal cancer cells. Neoplasia. 2010;12(8):618–627. doi: 10.1593/neo.10388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Hegde S, Kaushal N, Ravindra KC, et al. Δ12-prostaglandin J3, an omega-3 fatty acid–derived metabolite, selectively ablates leukemia stem cells in mice. Blood. 2011;118(26):6909–6919. doi: 10.1182/blood-2010-11-317750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Il Lee S, Zuo X, Shureiqi I. 15-Lipoxygenase-1 as a tumor suppressor gene in colon cancer: is the verdict in? Cancer Metastasis Rev. 2011;30(3–4):481–491. doi: 10.1007/s10555-011-9321-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Cathcart MC, Lysaght J, Pidgeon GP. Eicosanoid signalling pathways in the development and progression of colorectal cancer: novel approaches for prevention/intervention. Cancer Metastasis Rev. 2011;30(3–4):363–385. doi: 10.1007/s10555-011-9324-x. [DOI] [PubMed] [Google Scholar]
- 59.Cianchi F, Cortesini C, Magnelli L, et al. Inhibition of 5-lipoxygenase by MK886 augments the antitumor activity of celecoxib in human colon cancer cells. Mol Cancer Ther. 2006;5(11):2716–2726. doi: 10.1158/1535-7163.MCT-06-0318. [DOI] [PubMed] [Google Scholar]
- 60.Martel-Pelletier J, Lajeunesse D, Reboul P, Pelletier JP. Therapeutic role of dual inhibitors of 5-LOX and COX, selective and non-selective non-steroidal anti-inflammatory drugs. Ann Rheum Dis. 2003;62(6):501–509. doi: 10.1136/ard.62.6.501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Connor KM, SanGiovanni JP, Lofqvist C, et al. Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat Med. 2007;13(7):868–873. doi: 10.1038/nm1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Miller C, Yamaguchi RY, Ziboh VA. Guinea pig epidermis generates putative anti-inflammatory metabolites from fish oil polyunsaturated fatty acids. Lipids. 1989;24(12):998–1003. doi: 10.1007/BF02544068. [DOI] [PubMed] [Google Scholar]
- 63.Kim HY, Karanian JW, Salem N., Jr Formation of 15-lipoxygenase product from docosahexaenoic acid (22-6w3) by human platelets. Prostaglandins. 1990;40(5):539–549. doi: 10.1016/0090-6980(90)90115-c. [DOI] [PubMed] [Google Scholar]
- 64.Ziboh VA, Miller CC, Cho Y. Metabolism of polyunsaturated fatty acids by skin epidermal enzymes: generation of antiinflammatory and antiproliferative metabolites. Am J Clin Nutr. 2000;71(1 Suppl):361s–366s. doi: 10.1093/ajcn/71.1.361s. [DOI] [PubMed] [Google Scholar]
- 65.Bento AF, Claudino RF, Dutra RC, Marcon R, Calixto JB. Omega-3 fatty acid-derived mediators 17(R)-hydroxy docosahexaenoic acid, aspirin-triggered resolvin D1 and resolvin D2 prevent experimental colitis in mice. J Immunol. 2011;187(4):1957–1969. doi: 10.4049/jimmunol.1101305. [DOI] [PubMed] [Google Scholar]
- 66.Chiu CY, Gomolka B, Dierkes C, et al. Omega-6 docosapentaenoic acid-derived resolvins and 17-hydroxydocosahexaenoic acid modulate macrophage function and alleviate experimental colitis. Inflamm Res. 2012;61(9):967–976. doi: 10.1007/s00011-012-0489-8. [DOI] [PubMed] [Google Scholar]
- 67.Vang K, Ziboh VA. 15-lipoxygenase metabolites of gamma-linolenic acid/eicosapentaenoic acid suppress growth and arachidonic acid metabolism in human prostatic adenocarcinoma cells: possible implications of dietary fatty acids. Prostaglandins Leukot Essent Fatty Acids. 2005;72(5):363–372. doi: 10.1016/j.plefa.2005.02.002. [DOI] [PubMed] [Google Scholar]
- 68.O'Flaherty JT, Hu Y, Wooten RE, et al. 15-lipoxygenase metabolites of docosahexaenoic acid inhibit prostate cancer cell proliferation and survival. PLoS One. 2012;7(9):e45480. doi: 10.1371/journal.pone.0045480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hu Y, Sun H, O'Flaherty JT, Edwards IJ. 15-Lipoxygenase-1-mediated metabolism of docosahexaenoic acid is required for syndecan-1 signaling and apoptosis in prostate cancer cells. Carcinogenesis. 2013;34(1):176–182. doi: 10.1093/carcin/bgs324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Gleissman H, Yang R, Martinod K, et al. Docosahexaenoic acid metabolome in neural tumors: identification of cytotoxic intermediates. Faseb J. 2010;24(3):906–915. doi: 10.1096/fj.09-137919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Dwyer JH, Allayee H, Dwyer KM, et al. Arachidonate 5-lipoxygenase promoter genotype, dietary arachidonic acid, and atherosclerosis. N Engl J Med. 2004;350(1):29–37. doi: 10.1056/NEJMoa025079. [DOI] [PubMed] [Google Scholar]
- 72.Zhang G, Kodani S, Hammock BD. Stabilized epoxygenated fatty acids regulate inflammation, pain, angiogenesis and cancer. Prog Lipid Res. 2014;53(0):108–123. doi: 10.1016/j.plipres.2013.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kroetz DL, Xu F. Regulation and inhibition of arachidonic acid omegahydroxylases and 20-HETE formation. Annu Rev Pharmacol Toxicol. 2005;45:413–438. doi: 10.1146/annurev.pharmtox.45.120403.100045. [DOI] [PubMed] [Google Scholar]
- 74.Panigrahy D, Kalish BT, Huang S, et al. Epoxyeicosanoids promote organ and tissue regeneration. Proc Natl Acad Sci U S A. 2013;110(33):13528–13533. doi: 10.1073/pnas.1311565110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Panigrahy D, Edin ML, Lee CR, et al. Epoxyeicosanoids stimulate multiorgan metastasis and tumor dormancy escape in mice. J Clin Invest. 2012;122(1):178–191. doi: 10.1172/JCI58128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zhang W, Liao J, Li H, et al. Reduction of inflammatory bowel disease-induced tumor development in IL-10 knockout mice with soluble epoxide hydrolase gene deficiency. Mol Carcinog. 2013;52(9):726–738. doi: 10.1002/mc.21918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zhang W, Yang AL, Liao J, et al. Soluble epoxide hydrolase gene deficiency or inhibition attenuates chronic active inflammatory bowel disease in IL-10(−/−) mice. Dig Dis Sci. 2012;57(10):2580–2591. doi: 10.1007/s10620-012-2217-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Arnold C, Markovic M, Blossey K, et al. Arachidonic acid-metabolizing cytochrome P450 enzymes are targets of {omega}-3 fatty acids. J Biol Chem. 2010;285(43):32720–32733. doi: 10.1074/jbc.M110.118406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lucas D, Goulitquer S, Marienhagen J, et al. Stereoselective epoxidation of the last double bond of polyunsaturated fatty acids by human cytochromes P450. J Lipid Res. 2010;51(5):1125–1133. doi: 10.1194/jlr.M003061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Schwarz D, Kisselev P, Ericksen SS, et al. Arachidonic and eicosapentaenoic acid metabolism by human CYP1A1: highly stereoselective formation of 17(R),18(S)-epoxyeicosatetraenoic acid. Biochem Pharmacol. 2004;67(8):1445–1457. doi: 10.1016/j.bcp.2003.12.023. [DOI] [PubMed] [Google Scholar]
- 81.Barbosa-Sicard E, Markovic M, Honeck H, Christ B, Muller DN, Schunck WH. Eicosapentaenoic acid metabolism by cytochrome P450 enzymes of the CYP2C subfamily. Biochem Biophys Res Commun. 2005;329(4):1275–1281. doi: 10.1016/j.bbrc.2005.02.103. [DOI] [PubMed] [Google Scholar]
- 82.Morisseau C, Inceoglu B, Schmelzer K, et al. Naturally occurring monoepoxides of eicosapentaenoic acid and docosahexaenoic acid are bioactive antihyperalgesic lipids. J Lipid Res. 2010;51(12):3481–3490. doi: 10.1194/jlr.M006007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Shearer GC, Harris WS, Pedersen TL, Newman JW. Detection of omega-3 oxylipins in human plasma and response to treatment with omega-3 acid ethyl esters. J Lipid Res. 2010;51(8):2074–2081. doi: 10.1194/jlr.M900193-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Keenan AH, Pedersen TL, Fillaus K, Larson MK, Shearer GC, Newman JW. Basal omega-3 fatty acid status affects fatty acid and oxylipin responses to high-dose n3-HUFA in healthy volunteers. J Lipid Res. 2012;53(8):1662–1669. doi: 10.1194/jlr.P025577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zivkovic A, Yang J, Georgi K, et al. Serum oxylipin profiles in IgA nephropathy patients reflect kidney functional alterations. Metabolomics. 2012;8(6):1102–1113. doi: 10.1007/s11306-012-0417-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Schuchardt JP, Schmidt S, Kressel G, et al. Modulation of blood oxylipin levels by long-chain omega-3 fatty acid supplementation in hyper- and normolipidemic men. Prostaglandins Leukot Essent Fatty Acids. 2014;90(2–3):27–37. doi: 10.1016/j.plefa.2013.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Fischer R, Konkel A, Mehling H, et al. Dietary Omega-3 Fatty Acids Modulate the Eicosanoid Profile in Man Primarily via the CYP-epoxygenase Pathway. J Lipid Res. 2014 doi: 10.1194/jlr.M047357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Morin C, Sirois M, Echave V, Gomes MM, Rousseau E. EET displays anti-inflammatory effects in TNF-alpha stimulated human bronchi: putative role of CPI-17. Am J Respir Cell Mol Biol. 2008;38(2):192–201. doi: 10.1165/rcmb.2007-0232OC. [DOI] [PubMed] [Google Scholar]
- 89.Morin C, Sirois M, Echave V, Albadine R, Rousseau E. 17,18-epoxyeicosatetraenoic acid targets PPARgamma and p38 mitogen-activated protein kinase to mediate its anti-inflammatory effects in the lung: role of soluble epoxide hydrolase. Am J Respir Cell Mol Biol. 2010;43(5):564–575. doi: 10.1165/rcmb.2009-0155OC. [DOI] [PubMed] [Google Scholar]
- 90.Kubota T, Arita M, Isobe Y, et al. Eicosapentaenoic acid is converted via omega-3 epoxygenation to the anti-inflammatory metabolite 12-hydroxy-17,18-epoxyeicosatetraenoic acid. FASEB J. 2013 doi: 10.1096/fj.13-236224. [DOI] [PubMed] [Google Scholar]
- 91.Ye D, Zhang D, Oltman C, Dellsperger K, Lee HC, VanRollins M. Cytochrome p-450 epoxygenase metabolites of docosahexaenoate potently dilate coronary arterioles by activating large-conductance calcium-activated potassium channels. J Pharmacol Exp Ther. 2002;303(2):768–776. doi: 10.1124/jpet.303.2.768. [DOI] [PubMed] [Google Scholar]
- 92.Ulu A, Stephen Lee KS, Miyabe C, et al. An Omega-3 Epoxide of Docosahexaenoic Acid Lowers Blood Pressure in Angiotensin-II Dependent Hypertension. J Cardiovasc Pharmacol. 2014 doi: 10.1097/FJC.0000000000000094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Agbor LN, Walsh MT, Boberg JR, Walker MK. Elevated blood pressure in cytochrome P4501A1 knockout mice is associated with reduced vasodilation to omega-3 polyunsaturated fatty acids. Toxicol Appl Pharmacol. 2012;264(3):351–360. doi: 10.1016/j.taap.2012.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Cui PH, Petrovic N, Murray M. The omega-3 epoxide of eicosapentaenoic acid inhibits endothelial cell proliferation by p38 MAP kinase activation and cyclin D1/CDK4 downregulation. Br J Pharmacol. 2011;162(5):1143–1155. doi: 10.1111/j.1476-5381.2010.01113.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Chaffer CL, Weinberg RA. A Perspective on Cancer Cell Metastasis. Science. 2011;331(6024):1559–1564. doi: 10.1126/science.1203543. [DOI] [PubMed] [Google Scholar]
- 96.Hu J, Popp R, Fromel T, et al. Muller glia cells regulate Notch signaling and retinal angiogenesis via the generation of 19,20-dihydroxydocosapentaenoic acid. J Exp Med. 2014;211(2):281–295. doi: 10.1084/jem.20131494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Shao Z, Fu Z, Stahl A, et al. Cytochrome P450 2C8 omega3-long-chain polyunsaturated fatty acid metabolites increase mouse retinal pathologic neovascularization--brief report. Arterioscler Thromb Vasc Biol. 2014;34(3):581–586. doi: 10.1161/ATVBAHA.113.302927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Yang J, Schmelzer K, Georgi K, Hammock BD. Quantitative profiling method for oxylipin metabolome by liquid chromatography electrospray ionization tandem mass spectrometry. Anal Chem. 2009;81(19):8085–8093. doi: 10.1021/ac901282n. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Schmelzer K, Fahy E, Subramaniam S, Dennis EA. The lipid maps initiative in lipidomics. Methods Enzymol. 2007;432:171–183. doi: 10.1016/S0076-6879(07)32007-7. [DOI] [PubMed] [Google Scholar]
- 100.Zeisel SH, Waterland RA, Ordovas JM, Muoio DM, Jia W, Fodor A. Highlights of the 2012 Research Workshop: Using nutrigenomics and metabolomics in clinical nutrition research. JPEN J Parenter Enteral Nutr. 2013;37(2):190–200. doi: 10.1177/0148607112462401. [DOI] [PMC free article] [PubMed] [Google Scholar]



